Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Conformational strains of pathogenic amyloid proteins in neurodegenerative diseases

Abstract

Amyloid proteins, which are considered ‘villains’ in many neurodegenerative diseases, form enigmatic pathological strains that underlie disease pathogenesis and progression. Recent technical advances in cryogenic electron microscopy and solid-state NMR spectroscopy have enabled the high-resolution structures of full-length amyloid fibrils to be determined, initiating an era in which we have the opportunity to gain atomic-level structural understanding of pathogenic protein aggregation in neurodegenerative diseases. In this Review, we aim to explain the clinicopathological heterogeneity of neurodegenerative diseases by considering the polymorphic structures of amyloid fibrils. We decipher the structural basis for the generation of fibril polymorphs, how the fibril polymorphs differ in different disease contexts and how conformational changes alter the pathology caused by amyloid proteins during disease progression. Finally, we evaluate how this knowledge might aid clinical diagnostic and therapeutic strategies to treat neurodegenerative diseases.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Polymorphic strains of pathogenic proteins underlie neurodegenerative disease heterogeneity.
Fig. 2: Structural basis of amyloid fibril polymorphs.
Fig. 3: Changes in α-synuclein polymorphic fibrils in different contexts.
Fig. 4: A model of the pathological profiles of amyloid fibril polymorphs.
Fig. 5: Conformational transition of amyloid fibrils during seeded growth.

Similar content being viewed by others

References

  1. Dobson, C. M., Knowles, T. P. J. & Vendruscolo, M. The amyloid phenomenon and its significance in biology and medicine. Cold Spring Harb. Perspect. Biol. 12, a033878 (2019).

    Article  CAS  Google Scholar 

  2. Eisenberg, D. & Jucker, M. The amyloid state of proteins in human diseases. Cell 148, 1188–1203 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Peng, C., Trojanowski, J. Q. & Lee, V. M. Protein transmission in neurodegenerative disease. Nat. Rev. Neurol. 16, 199–212 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Milanesi, L. et al. Direct three-dimensional visualization of membrane disruption by amyloid fibrils. Proc. Natl Acad. Sci. USA 109, 20455–20460 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Engel, M. F. et al. Membrane damage by human islet amyloid polypeptide through fibril growth at the membrane. Proc. Natl Acad. Sci. USA 105, 6033–6038 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Stancu, I. C. et al. Aggregated tau activates NLRP3-ASC inflammasome exacerbating exogenously seeded and non-exogenously seeded tau pathology in vivo. Acta Neuropathol. 137, 599–617 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Meda, L. et al. Activation of microglial cells by beta-amyloid protein and interferon-gamma. Nature 374, 647–650 (1995).

    Article  CAS  PubMed  Google Scholar 

  8. Mahul-Mellier, A. L. et al. The process of Lewy body formation, rather than simply alpha-synuclein fibrillization, is one of the major drivers of neurodegeneration. Proc. Natl Acad. Sci. USA 117, 4971–4982 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Ruan, L. et al. Cytosolic proteostasis through importing of misfolded proteins into mitochondria. Nature 543, 443–446 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Olzscha, H. et al. Amyloid-like aggregates sequester numerous metastable proteins with essential cellular functions. Cell 144, 67–78 (2011).

    Article  CAS  PubMed  Google Scholar 

  11. Suzuki, G. et al. α-synuclein strains that cause distinct pathologies differentially inhibit proteasome. Elife 9, e56825 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Winslow, A. R. et al. α-Synuclein impairs macroautophagy: implications for Parkinson’s disease. J. Cell Biol. 190, 1023–1037 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Cuervo, A. M., Stefanis, L., Fredenburg, R., Lansbury, P. T. & Sulzer, D. Impaired degradation of mutant alpha-synuclein by chaperone-mediated autophagy. Science 305, 1292–1295 (2004).

    Article  CAS  PubMed  Google Scholar 

  14. Prusiner, S. B. Biology and genetics of prions causing neurodegeneration. Annu. Rev. Genet. 47, 601–623 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Jucker, M. & Walker, L. C. Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature 501, 45–51 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Luk, K. C. et al. Pathological alpha-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science 338, 949–953 (2012). This study reports that intrastriatal inoculation of α-syn fibrils causes cell-to-cell transmission and PD-like Lewy pathology in mouse brain. The PD mouse model developed in this study has been widely used for investigating the pathological properties of α-syn fibrils.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Prusiner, S. B. et al. Evidence for alpha-synuclein prions causing multiple system atrophy in humans with parkinsonism. Proc. Natl Acad. Sci. USA 112, E5308–E5317 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kaufman, S. K. et al. Tau prion strains dictate patterns of cell pathology, progression rate, and regional vulnerability in vivo. Neuron 92, 796–812 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kovacs, G. G. et al. Distribution patterns of tau pathology in progressive supranuclear palsy. Acta Neuropathol. 140, 99–119 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Kim, S. et al. Transneuronal propagation of pathologic alpha-synuclein from the gut to the brain models Parkinson’s disease. Neuron 103, 627–641 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Guo, J. L. et al. Distinct alpha-synuclein strains differentially promote tau inclusions in neurons. Cell 154, 103–117 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Braak, H. & Braak, E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 82, 239–259 (1991).

    Article  CAS  PubMed  Google Scholar 

  23. Braak, H. et al. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol. Aging 24, 197–211 (2003).

    Article  PubMed  Google Scholar 

  24. Vogel, J. W. et al. Four distinct trajectories of tau deposition identified in Alzheimer’s disease. Nat. Med. 27, 871–881 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Geddes, A. J., Parker, K. D., Atkins, E. D. & Beighton, E. “Cross-beta” conformation in proteins. J. Mol. Biol. 32, 343–358 (1968).

    Article  CAS  PubMed  Google Scholar 

  26. Jahn, T. R. et al. The common architecture of cross-beta amyloid. J. Mol. Biol. 395, 717–727 (2010).

    Article  CAS  PubMed  Google Scholar 

  27. Astbury, W. T., Dickinson, S. & Bailey, K. The X-ray interpretation of denaturation and the structure of the seed globulins. Biochem. J. 29, 2351–2360.1 (1935).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Qiang, W., Yau, W. M., Lu, J. X., Collinge, J. & Tycko, R. Structural variation in amyloid-beta fibrils from Alzheimer’s disease clinical subtypes. Nature 541, 217–221 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Falcon, B. et al. Structures of filaments from Pick’s disease reveal a novel tau protein fold. Nature 561, 137–140 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Gremer, L. et al. Fibril structure of amyloid-beta(1-42) by cryo-electron microscopy. Science 358, 116–119 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Fitzpatrick, A. W. P. et al. Cryo-EM structures of tau filaments from Alzheimer’s disease. Nature 547, 185–190 (2017). This study reveals, for the first time, the cryo-EM structures of tau amyloid fibrils directly extracted from the brains of patients with AD.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Li, Y. et al. Amyloid fibril structure of alpha-synuclein determined by cryo-electron microscopy. Cell Res. 28, 897–903 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Tuttle, M. D. et al. Solid-state NMR structure of a pathogenic fibril of full-length human alpha-synuclein. Nat. Struct. Mol. Biol. 23, 409–415 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Paravastu, A. K., Leapman, R. D., Yau, W. M. & Tycko, R. Molecular structural basis for polymorphism in Alzheimer’s beta-amyloid fibrils. Proc. Natl Acad. Sci. USA 105, 18349–18354 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Walti, M. A. et al. Atomic-resolution structure of a disease-relevant Abeta(1-42) amyloid fibril. Proc. Natl Acad. Sci. USA 113, E4976–E4984 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Radamaker, L. et al. Cryo-EM reveals structural breaks in a patient-derived amyloid fibril from systemic AL amyloidosis. Nat. Commun. 12, 875 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Shankar, G. M. et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat. Med. 14, 837–842 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kayed, R. et al. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science 300, 486–489 (2003).

    Article  CAS  PubMed  Google Scholar 

  39. Lashuel, H. A., Hartley, D., Petre, B. M., Walz, T. & Lansbury, P. T. Jr. Neurodegenerative disease: amyloid pores from pathogenic mutations. Nature 418, 291 (2002).

    Article  CAS  PubMed  Google Scholar 

  40. Winner, B. et al. In vivo demonstration that alpha-synuclein oligomers are toxic. Proc. Natl Acad. Sci. USA 108, 4194–4199 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Otzen, D. & Riek, R. Functional amyloids. Cold Spring Harb. Perspect. Biol. 11, a033860 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sawaya, M. R., Hughes, M. P., Rodriguez, J. A., Riek, R. & Eisenberg, D. S. The expanding amyloid family: structure, stability, function, and pathogenesis. Cell 184, 4857–4873 (2021).

    Article  CAS  PubMed  Google Scholar 

  43. Wei, G. et al. Self-assembling peptide and protein amyloids: from structure to tailored function in nanotechnology. Chem. Soc. Rev. 46, 4661–4708 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Hardy, J. A. & Higgins, G. A. Alzheimer’s disease: the amyloid cascade hypothesis. Science 256, 184–185 (1992).

    Article  CAS  PubMed  Google Scholar 

  45. Crowther, R. A. Straight and paired helical filaments in Alzheimer disease have a common structural unit. Proc. Natl Acad. Sci. USA 88, 2288–2292 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kidd, M. Paired helical filaments in electron microscopy of Alzheimer’s disease. Nature 197, 192–193 (1963).

    Article  CAS  PubMed  Google Scholar 

  47. Spillantini, M. G. et al. Alpha-synuclein in Lewy bodies. Nature 388, 839–840 (1997).

    Article  CAS  PubMed  Google Scholar 

  48. Polymeropoulos, M. H. et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science 276, 2045–2047 (1997).

    Article  CAS  PubMed  Google Scholar 

  49. Lashuel, H. A., Overk, C. R., Oueslati, A. & Masliah, E. The many faces of alpha-synuclein: from structure and toxicity to therapeutic target. Nat. Rev. Neurosci. 14, 38–48 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wetzel, R., Shivaprasad, S. & Williams, A. D. Plasticity of amyloid fibrils. Biochemistry 46, 1–10 (2007).

    Article  CAS  PubMed  Google Scholar 

  51. Benson, M. D. et al. Amyloid nomenclature 2018: recommendations by the International Society of Amyloidosis (ISA) nomenclature committee. Amyloid 25, 215–219 (2018).

    Article  CAS  PubMed  Google Scholar 

  52. Sunde, M. et al. Common core structure of amyloid fibrils by synchrotron X-ray diffraction. J. Mol. Biol. 273, 729–739 (1997).

    Article  CAS  PubMed  Google Scholar 

  53. Nelson, R. et al. Structure of the cross-beta spine of amyloid-like fibrils. Nature 435, 773–778 (2005). This study demonstrates the high structural diversity of amyloid-like fibril spines formed in vitro by amyloidogenic peptides at the atomic level.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Khurana, R. et al. Mechanism of thioflavin T binding to amyloid fibrils. J. Struct. Biol. 151, 229–238 (2005).

    Article  CAS  PubMed  Google Scholar 

  55. Collinge, J., Sidle, K. C., Meads, J., Ironside, J. & Hill, A. F. Molecular analysis of prion strain variation and the aetiology of ‘new variant’ CJD. Nature 383, 685–690 (1996).

    Article  CAS  PubMed  Google Scholar 

  56. Zhao, K. et al. Parkinson’s disease-related phosphorylation at Tyr39 rearranges alpha-synuclein amyloid fibril structure revealed by cryo-EM. Proc. Natl Acad. Sci. USA 117, 20305–20315 (2020). This study reveals the structural basis by which a PD-related phosphorylation determines the formation of a new fold within an α-syn fibril with enhanced neurotoxicity.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Zhao, K. et al. Parkinson’s disease associated mutation E46K of alpha-synuclein triggers the formation of a distinct fibril structure. Nat. Commun. 11, 2643 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Wang, L. Q. et al. Cryo-EM structure of an amyloid fibril formed by full-length human prion protein. Nat. Struct. Mol. Biol. 27, 598–602 (2020).

    Article  PubMed  CAS  Google Scholar 

  59. Sun, Y. et al. Cryo-EM structure of full-length alpha-synuclein amyloid fibril with Parkinson’s disease familial A53T mutation. Cell Res. 30, 360–362 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Guerrero-Ferreira, R. et al. Two new polymorphic structures of human full-length alpha-synuclein fibrils solved by cryo-electron microscopy. Elife 8, e48907 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Petkova, A. T. et al. Self-propagating, molecular-level polymorphism in Alzheimer’s beta-amyloid fibrils. Science 307, 262–265 (2005). This study reveals that Aβ can form amyloid fibrils with distinct molecular structures and neurotoxicity.

    Article  CAS  PubMed  Google Scholar 

  62. Petkova, A. T., Yau, W. M. & Tycko, R. Experimental constraints on quaternary structure in Alzheimer’s beta-amyloid fibrils. Biochemistry 45, 498–512 (2006).

    Article  CAS  PubMed  Google Scholar 

  63. Colletier, J. P. et al. Molecular basis for amyloid-beta polymorphism. Proc. Natl Acad. Sci. USA 108, 16938–16943 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Sawaya, M. R. et al. Atomic structures of amyloid cross-beta spines reveal varied steric zippers. Nature 447, 453–457 (2007).

    Article  CAS  PubMed  Google Scholar 

  65. Li, B. et al. Cryo-EM of full-length alpha-synuclein reveals fibril polymorphs with a common structural kernel. Nat. Commun. 9, 3609 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Guerrero-Ferreira, R. et al. Cryo-EM structure of alpha-synuclein fibrils. Elife 7, e36402 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Shi, Y. et al. Structure-based classification of tauopathies. Nature 598, 359–363 (2021). This study reports the structure of tau fibrils extracted from brain tissues of patients with progressive supranuclear palsy, and demonstrates that different tau fibril structures can be used for hierarchical classification of tauopathies.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Zhang, W. et al. Novel tau filament fold in corticobasal degeneration. Nature 580, 283–287 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Falcon, B. et al. Novel tau filament fold in chronic traumatic encephalopathy encloses hydrophobic molecules. Nature 568, 420–423 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Schweighauser, M. et al. Structures of alpha-synuclein filaments from multiple system atrophy. Nature 585, 464–469 (2020). This study reports, for the first time, the cryo-EM structures of α-syn fibrils extracted from patients’ brains, and shows the structural difference between in vitro-prepared α-syn fibrils and brain-extracted α-syn fibrils.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Boyer, D. R. et al. The alpha-synuclein hereditary mutation E46K unlocks a more stable, pathogenic fibril structure. Proc. Natl Acad. Sci. USA 117, 3592–3602 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Fersht, A. R. From the first protein structures to our current knowledge of protein folding: delights and scepticisms. Nat. Rev. Mol. Cell Biol. 9, 650–654 (2008).

    Article  CAS  PubMed  Google Scholar 

  73. Onuchic, J. N., Luthey-Schulten, Z. & Wolynes, P. G. Theory of protein folding: the energy landscape perspective. Annu. Rev. Phys. Chem. 48, 545–600 (1997).

    Article  CAS  PubMed  Google Scholar 

  74. Dill, K. A. & MacCallum, J. L. The protein-folding problem, 50 years on. Science 338, 1042–1046 (2012).

    Article  CAS  PubMed  Google Scholar 

  75. Tayeb-Fligelman, E. et al. The cytotoxic Staphylococcus aureus PSMalpha3 reveals a cross-alpha amyloid-like fibril. Science 355, 831–833 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Luo, F. et al. Atomic structures of FUS LC domain segments reveal bases for reversible amyloid fibril formation. Nat. Struct. Mol. Biol. 25, 341–346 (2018).

    Article  CAS  PubMed  Google Scholar 

  77. Hughes, M. P. et al. Atomic structures of low-complexity protein segments reveal kinked beta sheets that assemble networks. Science 359, 698–701 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Kant, R. et al. A structural analysis of amyloid polymorphism in disease: clues for selective vulnerability? Preprint at https://doi.org/10.1101/2021.03.01.433317 (2021).

  79. McKinley, M. P., Bolton, D. C. & Prusiner, S. B. A protease-resistant protein is a structural component of the scrapie prion. Cell 35, 57–62 (1983).

    Article  CAS  PubMed  Google Scholar 

  80. Safar, J., Roller, P. P., Gajdusek, D. C. & Gibbs, C. J. Jr. Thermal stability and conformational transitions of scrapie amyloid (prion) protein correlate with infectivity. Protein Sci. 2, 2206–2216 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Li, D. et al. Designed amyloid fibers as materials for selective carbon dioxide capture. Proc. Natl Acad. Sci. USA 111, 191–196 (2014).

    Article  CAS  PubMed  Google Scholar 

  82. Torrent, J. et al. High pressure induces scrapie-like prion protein misfolding and amyloid fibril formation. Biochemistry 43, 7162–7170 (2004).

    Article  CAS  PubMed  Google Scholar 

  83. Boyer, D. R. et al. Structures of fibrils formed by alpha-synuclein hereditary disease mutant H50Q reveal new polymorphs. Nat. Struct. Mol. Biol. 26, 1044–1052 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Arakhamia, T. et al. Posttranslational modifications mediate the structural diversity of tauopathy strains. Cell 180, 633–644 e612 (2020). This study reports the cryo-EM structure of tau fibrils extracted from the brains of patients with corticobasal degeneration, and demonstrates that PTMs are important in determining the structural polymorphs of tau fibril in different tauopathies.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Li, D. & Liu, C. Hierarchical chemical determination of amyloid polymorphs in neurodegenerative disease. Nat. Chem. Biol. 17, 237–245 (2021).

    Article  CAS  PubMed  Google Scholar 

  86. Miura, T., Suzuki, K., Kohata, N. & Takeuchi, H. Metal binding modes of Alzheimer’s amyloid beta-peptide in insoluble aggregates and soluble complexes. Biochemistry 39, 7024–7031 (2000).

    Article  CAS  PubMed  Google Scholar 

  87. Zarranz, J. J. et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann. Neurol. 55, 164–173 (2004).

    Article  CAS  PubMed  Google Scholar 

  88. Zhang, W. et al. Heparin-induced tau filaments are polymorphic and differ from those in Alzheimer’s and Pick’s diseases. Elife 8, e43584 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Lovestam, S. et al. Seeded assembly in vitro does not replicate the structures of alpha-synuclein filaments from multiple system atrophy. FEBS Open Bio 11, 999–1013 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Fan, Y. et al. Different structures and pathologies of alpha-synuclein fibrils derived from preclinical and postmortem patients of Parkinson’s disease. Preprint at https://doi.org/10.1101/2021.11.02.467019 (2021).

  91. Peng, C. et al. Cellular milieu imparts distinct pathological alpha-synuclein strains in alpha-synucleinopathies. Nature 557, 558–563 (2018). This study shows that different intracellular milieus of neurons and oligodendrocytes can induce different α-syn fibril strains with distinct pathology.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Sanders, D. W. et al. Distinct tau prion strains propagate in cells and mice and define different tauopathies. Neuron 82, 1271–1288 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Mao, X. et al. Pathological alpha-synuclein transmission initiated by binding lymphocyte-activation gene 3. Science 353, aah3374 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Goedert, M. Alzheimer’s and Parkinson’s diseases: the prion concept in relation to assembled Abeta, tau, and alpha-synuclein. Science 349, 1255555 (2015).

    Article  PubMed  CAS  Google Scholar 

  95. Zhang, S. et al. Mechanistic basis for receptor-mediated pathological alpha-synuclein fibril cell-to-cell transmission in Parkinson’s disease. Proc. Natl Acad. Sci. USA 118, e2011196118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Davis, A. A., Leyns, C. E. G. & Holtzman, D. M. Intercellular spread of protein aggregates in neurodegenerative disease. Annu. Rev. Cell Dev. Biol. 34, 545–568 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Fares, M. B., Jagannath, S. & Lashuel, H. A. Reverse engineering Lewy bodies: how far have we come and how far can we go? Nat. Rev. Neurosci. 22, 111–131 (2021).

    Article  CAS  PubMed  Google Scholar 

  98. Shahnawaz, M. et al. Discriminating alpha-synuclein strains in Parkinson’s disease and multiple system atrophy. Nature 578, 273–277 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Strohaker, T. et al. Structural heterogeneity of alpha-synuclein fibrils amplified from patient brain extracts. Nat. Commun. 10, 5535 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Long, H. et al. Wild-type alpha-synuclein inherits the structure and exacerbated neuropathology of E46K mutant fibril strain by cross-seeding. Proc. Natl Acad. Sci. USA 118, e2012435118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Brahmachari, S. et al. Activation of tyrosine kinase c-Abl contributes to alpha-synuclein-induced neurodegeneration. J. Clin. Invest. 126, 2970–2988 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  102. McGlinchey, R. P. & Lee, J. C. Cysteine cathepsins are essential in lysosomal degradation of alpha-synuclein. Proc. Natl Acad. Sci. USA 112, 9322–9327 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. McGlinchey, R. P. et al. C-terminal alpha-synuclein truncations are linked to cysteine cathepsin activity in Parkinson’s disease. J. Biol. Chem. 294, 9973–9984 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Moors, T. E. et al. The subcellular arrangement of alpha-synuclein proteoforms in the Parkinson’s disease brain as revealed by multicolor STED microscopy. Acta Neuropathol. 142, 423–448 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Sorrentino, Z. A. et al. Physiological C-terminal truncation of alpha-synuclein potentiates the prion-like formation of pathological inclusions. J. Biol. Chem. 293, 18914–18932 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Luk, K. C. et al. Molecular and Biological Compatibility with Host Alpha-Synuclein Influences Fibril Pathogenicity. Cell Rep. 16, 3373–3387 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Kollmer, M. et al. Cryo-EM structure and polymorphism of Abeta amyloid fibrils purified from Alzheimer’s brain tissue. Nat. Commun. 10, 4760 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. Lu, J. X. et al. Molecular structure of beta-amyloid fibrils in Alzheimer’s disease brain tissue. Cell 154, 1257–1268 (2013). This study shows that Aβ40 fibrils seeded from patients with AD with distinct clinical histories exhibit different molecular architectures.

    Article  CAS  PubMed  Google Scholar 

  109. Ghosh, U., Thurber, K. R., Yau, W. M. & Tycko, R. Molecular structure of a prevalent amyloid-beta fibril polymorph from Alzheimer’s disease brain tissue. Proc. Natl Acad. Sci. USA 118, e2023089118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Yang, Y. et al. Cryo-EM structures of amyloid-beta 42 filaments from human brains. Science 375, 167–172 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Shi, Y. et al. Cryo-EM structures of tau filaments from Alzheimer’s disease with PET ligand APN-1607. Acta Neuropathol. 141, 697–708 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Burger, D. et al. Cryo-EM structure of alpha-synuclein fibrils amplified by PMCA from PD and MSA patient brains. Preprint at https://doi.org/10.1101/2021.07.08.451588 (2021).

  113. Lövestam, S. et al. Assembly of recombinant tau into filaments identical to those of Alzheimer’s disease and chronic traumatic encephalopathy. Elife 11, e76494 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  114. Schütz, A. et al. Atomic-resolution three-dimensional structure of amyloid beta fibrils bearing the Osaka mutation. Angew. Chem. Int. Ed. 54, 331–335 (2015).

    Article  CAS  Google Scholar 

  115. Wesseling, H. et al. Tau PTM profiles identify patient heterogeneity and stages of Alzheimer’s disease. Cell 183, 1699–1713 (2020). This study demonstrates that tau proteins isolated from the brain tissues of patients with AD exhibit different PTM patterns at different stages of AD.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Morris, M. et al. Tau post-translational modifications in wild-type and human amyloid precursor protein transgenic mice. Nat. Neurosci. 18, 1183–1189 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Dujardin, S. et al. Tau molecular diversity contributes to clinical heterogeneity in Alzheimer’s disease. Nat. Med. 26, 1256–1263 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Glynn, C. et al. Cryo-EM structure of a human prion fibril with a hydrophobic, protease-resistant core. Nat. Struct. Mol. Biol. 27, 417–423 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Aoyagi, A. et al. Abeta and tau prion-like activities decline with longevity in Alzheimer’s disease brains. Sci. Transl. Med. 11, eaat8462 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Lucic, V., Rigort, A. & Baumeister, W. Cryo-electron tomography: the challenge of doing structural biology in situ. J. Cell Biol. 202, 407–419 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Trinkaus, V. A. et al. In situ architecture of neuronal alpha-synuclein inclusions. Nat. Commun. 12, 2110 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Bauerlein, F. J. B., Fernandez-Busnadiego, R. & Baumeister, W. Investigating the structure of neurotoxic protein aggregates inside cells. Trends Cell Biol. 30, 951–966 (2020).

    Article  PubMed  CAS  Google Scholar 

  123. Irwin, D. J. et al. Deep clinical and neuropathological phenotyping of Pick disease. Ann. Neurol. 79, 272–287 (2016).

    Article  CAS  PubMed  Google Scholar 

  124. Montenigro, P. H., Corp, D. T., Stein, T. D., Cantu, R. C. & Stern, R. A. Chronic traumatic encephalopathy: historical origins and current perspective. Annu. Rev. Clin. Psychol. 11, 309–330 (2015).

    Article  PubMed  Google Scholar 

  125. Crary, J. F. et al. Primary age-related tauopathy (PART): a common pathology associated with human aging. Acta Neuropathol. 128, 755–766 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Forrest, S. L., Kril, J. J. & Halliday, G. M. Cellular and regional vulnerability in frontotemporal tauopathies. Acta Neuropathol. 138, 705–727 (2019).

    Article  PubMed  Google Scholar 

  127. Gotz, J., Halliday, G. & Nisbet, R. M. Molecular pathogenesis of the tauopathies. Annu. Rev. Pathol. 14, 239–261 (2019).

    Article  CAS  PubMed  Google Scholar 

  128. Saito, Y. et al. Staging of argyrophilic grains: an age-associated tauopathy. J. Neuropathol. Exp. Neurol. 63, 911–918 (2004).

    Article  PubMed  Google Scholar 

  129. Botez, G., Probst, A., Ipsen, S. & Tolnay, M. Astrocytes expressing hyperphosphorylated tau protein without glial fibrillary tangles in argyrophilic grain disease. Acta Neuropathol. 98, 251–256 (1999).

    Article  CAS  PubMed  Google Scholar 

  130. Kovacs, G. G. et al. Aging-related tau astrogliopathy (ARTAG): harmonized evaluation strategy. Acta Neuropathol. 131, 87–102 (2016).

    Article  CAS  PubMed  Google Scholar 

  131. Braak, H., Sastre, M. & Del Tredici, K. Development of alpha-synuclein immunoreactive astrocytes in the forebrain parallels stages of intraneuronal pathology in sporadic Parkinson’s disease. Acta Neuropathol. 114, 231–241 (2007).

    Article  CAS  PubMed  Google Scholar 

  132. Wakabayashi, K., Hayashi, S., Yoshimoto, M., Kudo, H. & Takahashi, H. NACP/alpha-synuclein-positive filamentous inclusions in astrocytes and oligodendrocytes of Parkinson’s disease brains. Acta Neuropathol. 99, 14–20 (2000).

    Article  CAS  PubMed  Google Scholar 

  133. Halliday, G. M., Holton, J. L., Revesz, T. & Dickson, D. W. Neuropathology underlying clinical variability in patients with synucleinopathies. Acta Neuropathol. 122, 187–204 (2011).

    Article  CAS  PubMed  Google Scholar 

  134. Terada, S. et al. Glial involvement in diffuse Lewy body disease. Acta Neuropathol. 105, 163–169 (2003).

    Article  CAS  PubMed  Google Scholar 

  135. Jellinger, K. A. Lewy body/alpha-synucleinopathy in schizophrenia and depression: a preliminary neuropathological study. Acta Neuropathol. 117, 423–427 (2009).

    Article  CAS  PubMed  Google Scholar 

  136. Jellinger, K. A. Lewy body-related alpha-synucleinopathy in the aged human brain. J. Neural Transm. 111, 1219–1235 (2004).

    Article  CAS  PubMed  Google Scholar 

  137. Shishido, T. et al. alpha-synuclein accumulation in skin nerve fibers revealed by skin biopsy in pure autonomic failure. Neurology 74, 608–610 (2010).

    Article  CAS  PubMed  Google Scholar 

  138. Kaufmann, H., Hague, K. & Perl, D. Accumulation of alpha-synuclein in autonomic nerves in pure autonomic failure. Neurology 56, 980–981 (2001).

    Article  CAS  PubMed  Google Scholar 

  139. Coon, E. A., Singer, W. & Low, P. A. Pure autonomic failure. Mayo Clin. Proc. 94, 2087–2098 (2019).

    Article  PubMed  Google Scholar 

  140. Kovari, E., Burkhardt, K., Lobrinus, J. A. & Bouras, C. Lewy body dysphagia. Acta Neuropathol. 114, 295–298 (2007).

    Article  PubMed  Google Scholar 

  141. Jackson, M., Lennox, G., Balsitis, M. & Lowe, J. Lewy body dysphagia. J. Neurol. Neurosurg. Psychiatry 58, 756–758 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Nakamura, K. et al. Accumulation of phosphorylated alpha-synuclein in subpial and periventricular astrocytes in multiple system atrophy of long duration. Neuropathology 36, 157–167 (2016).

    Article  CAS  PubMed  Google Scholar 

  143. Vabulas, M. et al. Protein folding in the cytoplasm and the heat shock response. Cold Spring Harb. Perspect. Biol. 2, a004390 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank Y. Ma, Y. Fan and H. Long for assisting with the preparation of figures and tables. They also thank the National Natural Science Foundation of China (grants 82188101, 32170683, 31872716 and 32171236), the Major State Basic Research Development Program (grant 2019YFE0120600), the Science and Technology Commission of Shanghai Municipality (grants 20XD1425000 and 2019SHZDZX02), the Chinese Academy of Science Project for Young Scientists in Basic Research (grant YSBR-009) and the Shanghai Pilot Program for Basic Research–Chinese Academy of Science, Shanghai Branch (grant CYJ-SHFY-2022-005), for funding support.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding authors

Correspondence to Dan Li or Cong Liu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Neuroscience thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

RCSB Protein Data Bank: https://www.rcsb.org/

Glossary

Amyloid fibrils

Insoluble aggregates, formed by repetitive supermolecular assembly of a misfolded protein, in which the protein units are ordered in β-strand-rich structures.

Self-propagation

Process in which preformed amyloid fibrils of a protein nucleate the conversion of the soluble form of the protein into the amyloid form to generate more fibrils.

Cross-β structure

A common structural feature of amyloid fibrils that arises from β-sheet bundles in which the direction of β-strands is nearly perpendicular to the fibril axis.

Cryogenic electron microscopy

(cryo-EM). A transmission electron microscopy technique that is applied to samples that have been rapidly frozen into a glass-like state.

Solid-state NMR spectroscopy

An NMR technology used to investigate the chemical structure and dynamics of solids and semi-solids at an atomic level.

Protein strains

Proteins that form the unique fibril structures that underlie the pathological presentation of a disease.

Conformational strains

Forms of a protein with distinct fibril structures.

Prion diseases

A family of neurodegenerative disorders caused by misfolded prion protein that affect both humans and animals.

Isoforms

Different forms of a protein produced from alternative splicing of the same gene or from similar genes.

Fibril polymorphs

Amyloid fibrils with different structures formed from the same protein.

Post-translational modifications

(PTMs). Biochemical modifications of one or more amino acids within a protein that occur after translation.

Protein misfolding

A common cellular event in which a protein fails to achieve its native structure, resulting in functional abnormality.

Intrinsically disordered proteins

Proteins that lack unique 3D structures but may transit to ordered physiological structures upon interaction with binding partners or to pathological structures under disease conditions.

α-Helix

A type of secondary structure in which the protein chain is coiled as a result of regularly spaced hydrogen bonding between residues and in which the side chains are left reaching outwards.

β-Strand

A type of secondary structure in which the protein chain is extended as a result of the backbone forming hydrogen bonds with the adjacent chains and in which the side chains alternate on both sides.

Free energy

Also called Gibbs free energy. A thermodynamic quantity that expresses the amount of work that can be done by a system. In protein folding, the change of free energy is used to describe the thermodynamic stability of a protein.

Energy landscape

A model that describes protein folding as a funnel-like landscape gradually biased towards the energetically optimal structure.

Cofactors

Substances essential for the construction of the fibril.

Charged pockets

Cavities on the surface or in the interior of a fibril that contain charged residues.

Salt bridges

Electrostatic attractions between oppositely charged amino acid side chains.

β-Hairpin motif

A protein structural motif in which the adjacent antiparallel β-strands are connected either by backbone hydrogen bonding (as usually seen in native structures) or by side chain interactions (as seen in fibril structures).

Seeds

Preformed amyloid fibrils that can serve as a nucleus to template and accelerate the conversion of soluble proteins to fibrils.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, D., Liu, C. Conformational strains of pathogenic amyloid proteins in neurodegenerative diseases. Nat Rev Neurosci 23, 523–534 (2022). https://doi.org/10.1038/s41583-022-00603-7

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41583-022-00603-7

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing